Recombinant Chicken Myotubularin-related protein 2 (MTMR2), partial

Shipped with Ice Packs
In Stock

Description

Definition and Production Systems

Recombinant Chicken MTMR2 (partial) refers to a truncated version of the full-length protein produced using heterologous expression systems. Commercial providers like CUSABIO offer this protein in multiple formats:

Product CodeExpression SystemTag/Modification
CSB-YP719990CHYeastNative
CSB-EP719990CHE. coliStandard purification
CSB-BP719990CHBaculovirus-insect cellsHigh-yield production
CSB-MP719990CHMammalian cellsPost-translational modifications
CSB-EP719990CH-BE. coliIn vivo biotinylation

These systems enable researchers to study MTMR2's enzymatic activity, structural properties, and interactions in vitro .

Phosphoinositide Regulation

MTMR2 orthologs dephosphorylate PI(3)P and PI(3,5)P₂, lipids enriched in endosomal membranes. This activity modulates:

  • Endosomal maturation and signaling .

  • Mechanotransduction via Piezo2 ion channel regulation .

  • Myelin maintenance in peripheral nerves .

Disease Relevance

Mutations in human MTMR2 cause Charcot-Marie-Tooth disease type 4B1, a neuropathy characterized by myelin abnormalities . While chicken MTMR2 has not been directly linked to disease, its conserved domains suggest analogous roles in membrane trafficking and signaling.

Research Applications

Recombinant Chicken MTMR2 (partial) is primarily used for:

  • Enzymatic assays: Measuring phosphatase activity against PI(3)P/PI(3,5)P₂ substrates.

  • Protein interaction studies: Mapping binding partners like APPL1 or Rab5 effectors .

  • Structural analysis: Crystallography or NMR to resolve domain architectures .

Limitations and Knowledge Gaps

  • Sequence coverage: The "partial" designation implies missing regions (e.g., C-terminal PDZ-binding motifs), which may alter function .

  • Species-specific differences: Chicken MTMR2 may exhibit unique regulatory features compared to mammalian orthologs.

  • Validation data: Direct studies on chicken MTMR2’s substrate specificity or cellular roles are absent in published literature.

Future Directions

Key unanswered questions include:

  • Does phosphorylation at residues analogous to human Ser58/S631 regulate chicken MTMR2’s endosomal shuttling?

  • Can chicken MTMR2 compensate for human MTMR2 loss in disease models, as seen with MTM1/MTMR2 cross-rescue ?

  • How does its lipid phosphatase activity compare to human MTMR2 under stress conditions (e.g., osmotic shock) ?

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
MTMR2; RCJMB04_23g22; Myotubularin-related protein 2; Phosphatidylinositol-3,5-bisphosphate 3-phosphatase; EC 3.1.3.95; Phosphatidylinositol-3-phosphate phosphatase; EC 3.1.3.64
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Gallus gallus (Chicken)
Target Names
Uniprot No.

Target Background

Function
Phosphatase acting on lipids with phosphoinositol headgroups. Exhibits phosphatase activity towards phosphatidylinositol 3-phosphate and phosphatidylinositol 3,5-bisphosphate.
Database Links
Protein Families
Protein-tyrosine phosphatase family, Non-receptor class myotubularin subfamily
Subcellular Location
Cytoplasm. Early endosome membrane; Peripheral membrane protein.

Q&A

What is Myotubularin-related protein 2 and what are its primary functions in chickens?

Myotubularin-related protein 2 (MTMR2) belongs to the myotubularin family of phosphoinositide phosphatases. It functions as a 3-phosphatase with specificity for phosphoinositides PI(3)P and PI(3,5)P2, which are primarily located on endosomal membranes. While most research has focused on mammalian MTMR2, chicken MTMR2 likely serves similar fundamental roles in regulating endosomal trafficking, membrane dynamics, and signaling pathways.

In mammals, MTMR2 is crucial for proper neuronal function, as it localizes to excitatory synapses through direct interaction with postsynaptic scaffolding proteins like PSD-95. Knockdown of MTMR2 in cultured neurons significantly reduces excitatory synapse density and function, which can only be rescued by catalytically active MTMR2 that retains binding capacity to scaffolding proteins . Similar synaptic regulation mechanisms likely exist in the avian nervous system.

Additionally, MTMR2 plays important roles in mechanosensation by regulating Piezo2 ion channels, which are critical determinants of light touch sensation. Research demonstrates that MTMR2 attenuates Piezo2-mediated rapidly adapting mechanically activated (RA-MA) currents through local modulation of PI(3,5)P2 levels .

How does chicken MTMR2 structurally and functionally compare to mammalian orthologs?

While specific structural comparisons between chicken and mammalian MTMR2 have limited documentation in current literature, functional analyses can be extrapolated based on conserved domains and catalytic mechanisms. Mammalian MTMR2 contains a PH-GRAM domain important for membrane association and a coiled-coil dimerization module that influences its subcellular localization . The protein also contains a conserved C-terminal PDZ-binding motif that facilitates interaction with synaptic scaffolding proteins like PSD-95 .

The catalytic phosphatase domain of MTMR2 contains a conserved CX5R motif characteristic of the protein tyrosine phosphatase family, with the critical cysteine residue (C417 in mammalian MTMR2) being essential for phosphatase activity. Mutation of this residue results in a catalytically inactive form (C417S) . When designing recombinant chicken MTMR2 constructs, researchers should identify the corresponding catalytic residues through sequence alignment.

Mammalian MTMR2 expression gradually increases during postnatal brain development, with patterns similar to synaptic proteins like PSD-95 . Subcellular fractionation studies show enrichment in synaptic fractions, including crude synaptosomal (P2) and synaptosomal membrane (LP1) fractions, with detectable levels in postsynaptic density fractions (PSD I and PSD II) .

What approaches are recommended for sequence verification of recombinant chicken MTMR2?

For sequence verification of recombinant chicken MTMR2, a comprehensive approach combining multiple techniques is recommended:

  • PCR amplification and sequencing: Design primers targeting conserved regions based on published chicken MTMR2 sequences. Use high-fidelity DNA polymerase for amplification followed by Sanger sequencing of the product.

  • Multiple sequence alignment: Compare the obtained sequence with established MTMR2 sequences from other species, particularly focusing on catalytic domains and functional motifs. Key regions to verify include:

    • The catalytic CX5R motif in the phosphatase domain

    • The PH-GRAM domain

    • The C-terminal PDZ-binding motif

    • Phosphorylation sites (equivalent to mammalian Ser58 and Ser631)

  • Mass spectrometry validation: For recombinant protein validation, use LC-MS/MS to confirm protein identity and potential post-translational modifications. This is particularly important for verifying the correct expression of partial MTMR2 constructs.

  • Functional domain verification: Express the partial recombinant protein and conduct phosphatase activity assays using PI(3)P and PI(3,5)P2 substrates to confirm catalytic activity, which serves as functional verification of proper folding and domain structure.

For partial MTMR2 constructs, it's crucial to maintain intact catalytic domains while carefully documenting which regions are included or excluded in the recombinant protein.

What expression systems are most effective for producing recombinant chicken MTMR2?

The choice of expression system for recombinant chicken MTMR2 depends on experimental requirements for protein yield, post-translational modifications, and downstream applications. Based on approaches used for mammalian MTMR2 research, the following systems can be considered:

Bacterial expression (E. coli):

  • Advantages: High yield, cost-effective, rapid expression

  • Considerations: May lack post-translational modifications; potential issues with protein folding and solubility

  • Recommended for: Structural studies requiring large amounts of protein or domains without critical modifications

  • Expression vectors: pET series with N-terminal His6 or GST tags for purification

  • Strains: BL21(DE3), Rosetta(DE3) for rare codon optimization

Mammalian cell expression (HEK293T):

  • Advantages: Proper post-translational modifications, particularly phosphorylation at key regulatory sites (e.g., Ser58, Ser631); appropriate for functional studies

  • Considerations: Lower yield compared to bacterial systems

  • Recommended for: Functional assays, protein-protein interaction studies, subcellular localization experiments

  • Expression vectors: p3XFLAG-CMV7.1 has been successfully used for MTMR2 expression

Baculovirus-infected insect cells:

  • Advantages: Higher yield than mammalian systems while maintaining most post-translational modifications

  • Considerations: More complex setup than bacterial systems

  • Recommended for: Production of larger amounts of functionally active protein

When expressing partial MTMR2 constructs, careful design is essential to ensure proper folding and retention of catalytic activity if functional protein is desired.

What are the optimal purification strategies for recombinant chicken MTMR2?

Purification of recombinant chicken MTMR2 requires a multi-step approach to achieve high purity while maintaining enzyme activity. Based on established protocols for phosphoinositide phosphatases, the following strategy is recommended:

Step 1: Affinity chromatography

  • For His-tagged constructs: Ni-NTA affinity chromatography

    • Binding buffer: 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole

    • Washing buffer: Same as binding with increased imidazole (20-40 mM)

    • Elution buffer: Same as binding with 250-300 mM imidazole

  • For GST-tagged constructs: Glutathione sepharose chromatography

    • Binding buffer: PBS (pH 7.4)

    • Elution buffer: 50 mM Tris-HCl pH 8.0, 10 mM reduced glutathione

Step 2: Ion exchange chromatography

  • Recommended for removing contaminants with different charge properties

  • Use Q Sepharose for anion exchange or SP Sepharose for cation exchange depending on the theoretical pI of the protein

Step 3: Size exclusion chromatography

  • Final polishing step using Superdex 200 or similar

  • Running buffer: 20 mM HEPES pH 7.4, 150 mM NaCl, 1 mM DTT

Critical considerations:

  • Include protease inhibitors in all buffers to prevent degradation

  • Maintain reducing conditions (1-5 mM DTT or 2-10 mM β-mercaptoethanol) to protect the catalytic cysteine residue

  • For membrane-associated domains (e.g., PH-GRAM), consider adding 5-10% glycerol to all buffers to improve stability

  • Keep samples at 4°C throughout purification to minimize degradation

  • If phosphatase activity is required, avoid phosphate buffers which can inhibit enzyme activity

For partial MTMR2 constructs, adjust purification conditions based on the specific domains included and their biochemical properties.

How can I assess the stability and activity of purified recombinant chicken MTMR2?

A comprehensive assessment of purified recombinant chicken MTMR2 stability and activity should include:

Stability assessment:

  • Thermal shift assay (Differential Scanning Fluorimetry)

    • Monitors protein unfolding in response to temperature increase

    • Provides melting temperature (Tm) as a stability indicator

    • Can identify buffer conditions that enhance stability

  • Storage stability testing

    • Test protein activity retention after storage at different temperatures (-80°C, -20°C, 4°C)

    • Evaluate effect of freeze-thaw cycles on activity

    • Recommended storage conditions: 20 mM HEPES pH 7.4, 150 mM NaCl, 1 mM DTT, 10% glycerol

  • Size exclusion chromatography

    • Monitor for aggregation or degradation over time

    • Can detect oligomerization state changes

Activity assessment:

  • Malachite green phosphate release assay

    • Quantifies released phosphate from PI(3)P or PI(3,5)P2 substrates

    • Reaction buffer: 50 mM sodium acetate, pH 6.0, 25 mM KCl, 2 mM DTT

    • Incubate enzyme with substrate (typically 100 μM) at 37°C

    • Measure released phosphate using malachite green reagent

  • Thin-layer chromatography (TLC)

    • Monitors conversion of radiolabeled substrates to products

    • Provides direct visualization of phosphoinositide conversion

  • HPLC-based assay

    • Allows precise quantification of substrate and product

    • Suitable for detailed kinetic analysis

For partial MTMR2 constructs, activity measurement protocols should be adjusted based on which functional domains are present. If the partial construct contains the full catalytic domain, standard phosphatase assays can be applied. If regulatory domains are missing, activity may differ from the full-length protein, which should be documented and considered in experimental interpretation.

How do phosphorylation sites regulate chicken MTMR2 localization and function?

Phosphorylation plays a critical role in regulating MTMR2 subcellular localization and function. Based on mammalian MTMR2 studies, chicken MTMR2 likely has similar regulatory phosphorylation sites. Two key phosphorylation sites have been identified in mammalian MTMR2 that significantly impact its function:

Ser58 phosphorylation:

  • When phosphorylated, MTMR2 is sequestered in the cytoplasm

  • Dephosphorylation of Ser58 (S58A mutant) targets MTMR2 to Rab5-positive endosomes

  • This targeting results in PI(3)P depletion on endosomes and increased endosomal signaling, including enhanced ERK1/2 activation

  • Ser58 is phosphorylated by ERK1/2 in a negative feedback mechanism that regulates MTMR2 endosomal targeting

Ser631 phosphorylation:

  • Works in combination with Ser58 phosphorylation status

  • Regulates MTMR2 shuttling between different endosomal subtypes

  • While Ser58 phosphorylation status determines general endosomal binding, Ser631 phosphorylation mediates shuttling between Rab5-positive and APPL1-positive endosomal subtypes

  • A double phosphorylation-deficient mutant (S58A/S631A) shifts MTMR2 localization to APPL1-positive endosomes and leads to more sustained and pronounced ERK1/2 activation compared to the single S58A mutant

This differential phosphorylation creates a dynamic regulatory system that controls MTMR2 compartmentalization, influencing endosome maturation and signaling outcomes. For chicken MTMR2, researchers should identify the equivalent phosphorylation sites through sequence alignment and conduct site-directed mutagenesis studies to confirm similar regulatory mechanisms.

The following table summarizes the effects of different phosphorylation states:

Phosphorylation StateSubcellular LocalizationFunctional Effect
Ser58(+)/Ser631(+)CytoplasmicMinimal effect on endosomal PI(3)P, baseline ERK1/2 activation
Ser58(-)/Ser631(+)Rab5-positive endosomesReduced endosomal PI(3)P, increased ERK1/2 activation
Ser58(-)/Ser631(-)APPL1-positive endosomesSustained and pronounced increase in ERK1/2 activation
Ser58(+)/Ser631(-)Primarily cytoplasmicSimilar to double positive phosphorylation state

What experimental approaches can determine chicken MTMR2 substrate specificity?

Determining substrate specificity of chicken MTMR2 requires multiple complementary approaches:

In vitro enzymatic assays:

  • Phosphate release assays

    • Incubate purified MTMR2 with different potential substrates (PI(3)P, PI(3,5)P2, PI(5)P, etc.)

    • Measure released phosphate using malachite green or BIOMOL Green assays

    • Calculate kinetic parameters (Km, Vmax, kcat) for each substrate to determine preference

  • Radiolabeled substrate conversion

    • Use 32P-labeled phosphoinositides to directly monitor conversion

    • Analyze products by thin-layer chromatography

    • Quantify substrate-to-product conversion ratios for different phosphoinositides

  • Mass spectrometry-based lipidomics

    • Incubate MTMR2 with mixed phosphoinositide substrates

    • Extract and analyze lipids by LC-MS/MS

    • Provides comprehensive analysis of all substrate-product relationships

Cellular approaches:

  • Phosphoinositide-specific biosensors

    • Express fluorescent PI(3)P sensors (e.g., 2xFYVE-GFP) and PI(3,5)P2 sensors (e.g., ML1N-GFP)

    • Co-express wild-type or catalytically inactive MTMR2

    • Monitor changes in biosensor localization and intensity by confocal microscopy

  • Immunofluorescence with phosphoinositide-specific antibodies

    • Overexpress or knockdown MTMR2 in cultured cells

    • Fix and stain with antibodies specific for different phosphoinositides

    • Quantify changes in staining intensity and localization

  • Lipidomic analysis of endosomal fractions

    • Isolate endosomal fractions from cells with manipulated MTMR2 expression

    • Perform targeted lipidomic analysis to quantify changes in phosphoinositide levels

Structure-function approaches:

  • Mutation of catalytic site residues

    • Create point mutations in the catalytic pocket to alter substrate recognition

    • Compare activity against different substrates

    • Identify residues that determine substrate specificity

The catalytic activity of mammalian MTMR2 is highly dependent on the conserved cysteine residue in the CX5R motif. The C417S mutation abolishes phosphatase activity , and similar mutation in chicken MTMR2 would be expected to produce a catalytically inactive protein that can serve as an important negative control in substrate specificity studies.

How does chicken MTMR2 interact with the Piezo2 mechanosensory channel?

MTMR2 has been identified as an interactor with Piezo2, a mechanosensitive ion channel crucial for light touch sensation. The interaction between MTMR2 and Piezo2 has been demonstrated in mammalian systems, and similar mechanisms likely exist in chickens due to evolutionary conservation of these proteins' functions.

Key aspects of the MTMR2-Piezo2 interaction:

  • Physical proximity and interaction

    • Proximity ligation assays (PLA) demonstrate close proximity of MTMR2 and Piezo2 in both neuronal somata and neurites

    • MTMR2 was identified in the native Piezo2 interactome in dorsal root ganglia (DRG) with high enrichment (log2 9.96) and significance (p=0.00030)

  • Functional effects

    • MTMR2 attenuates Piezo2-mediated rapidly adapting mechanically activated (RA-MA) currents

    • This modulation appears specific to Piezo2, as Piezo1 and other MA current subtypes in DRG neurons were largely unaffected by MTMR2

    • Knockdown of MTMR2 potentiates Piezo2 RA-MA currents, while overexpression suppresses them

  • Mechanism of modulation

    • MTMR2 regulation of Piezo2 involves depletion of PI(3,5)P2

    • A specific PI(3,5)P2 binding region has been identified in Piezo2 (but not Piezo1) that confers sensitivity to MTMR2

    • Domain-swapping experiments confirm that this region is responsible for the differential effect of MTMR2 on Piezo2 versus Piezo1

Experimental approaches to study chicken MTMR2-Piezo2 interaction:

  • Co-immunoprecipitation assays

    • Express tagged versions of chicken MTMR2 and Piezo2 in heterologous systems

    • Perform pull-down experiments to confirm direct physical interaction

    • Use deletion constructs to map interaction domains

  • Proximity ligation assay (PLA)

    • Use primary antibodies against MTMR2 and Piezo2 in chicken DRG cultures

    • Quantify PLA signals in neuronal somata and neurites to confirm close proximity

    • Compare wild-type versus catalytically inactive MTMR2 to determine if enzymatic activity affects interaction

  • Electrophysiological assessment

    • Perform patch-clamp recordings of mechanically activated currents in chicken DRG neurons

    • Manipulate MTMR2 levels through overexpression or knockdown

    • Measure changes in Piezo2 RA-MA current amplitude, activation threshold, and inactivation kinetics

  • Phosphoinositide manipulation experiments

    • Use pharmacological inhibitors of PI(3,5)P2 synthesis in conjunction with MTMR2 manipulation

    • Employ osmotic stress to alter membrane tension and phosphoinositide distribution

    • Test effects on Piezo2 function to confirm PI(3,5)P2-dependent mechanisms

This interaction represents an important regulatory mechanism for mechanosensation and offers potential therapeutic targets for touch-related sensory disorders.

What approaches can identify novel binding partners of chicken MTMR2?

Identifying novel binding partners of chicken MTMR2 requires a multi-faceted approach combining unbiased screening methods with targeted validation techniques:

Unbiased screening methods:

  • Yeast two-hybrid screening

    • Use chicken MTMR2 or specific domains (e.g., C-terminal region, PH-GRAM domain) as bait

    • Screen against chicken cDNA libraries (preferably tissue-specific, such as brain or DRG)

    • This approach has successfully identified interactions between MTMR2 and PDZ domain-containing proteins

    • For partial MTMR2 constructs, ensure the domains of interest are properly expressed as bait

  • Proximity-dependent biotin identification (BioID)

    • Fuse chicken MTMR2 to a promiscuous biotin ligase (BirA*)

    • Express in relevant cell types (e.g., chicken DRG neurons or heterologous systems)

    • Identify biotinylated proteins in proximity to MTMR2 by streptavidin pull-down and mass spectrometry

  • Co-immunoprecipitation coupled with mass spectrometry

    • Express tagged chicken MTMR2 in relevant cell types

    • Perform immunoprecipitation under varied detergent conditions to preserve different interaction strengths

    • Identify co-precipitated proteins by mass spectrometry

    • Use catalytically inactive mutants (e.g., C417S equivalent) to identify interactions stabilized by substrate trapping

Validation techniques:

  • Reciprocal co-immunoprecipitation

    • Confirm interactions by pulling down with antibodies against candidate interactors

    • Western blot for MTMR2 in the precipitated complex

  • Domain mapping

    • Generate deletion constructs of MTMR2 to identify interaction domains

    • For key interactions, perform point mutations to identify critical residues

    • Previous work has shown that the C-terminal region (aa 637-643) of MTMR2 is critical for binding PDZ domain-containing proteins

  • Proximity ligation assay (PLA)

    • Visualize protein interactions in situ in fixed cells or tissues

    • Quantify interaction signals in different subcellular compartments

  • Functional validation

    • Assess how candidate interactors affect MTMR2 localization, activity, or stability

    • Evaluate how MTMR2 influences the function of interacting proteins

Considerations for partial MTMR2 constructs:

  • Carefully document which domains are present in the partial construct

  • Consider that some interactions may be lost if key binding regions are absent

  • Domain-specific interactors may be enriched when using focused constructs

  • Compare interactome results with full-length MTMR2 to identify domain-specific interactions

How can chicken MTMR2 be used to study endosomal trafficking and maturation?

Chicken MTMR2 provides a valuable tool for studying endosomal trafficking and maturation due to its role as a phosphoinositide phosphatase that regulates endosomal phosphoinositide composition. The following approaches can leverage MTMR2 for detailed endosomal studies:

Visualization and quantification approaches:

  • Live-cell imaging with phosphoinositide biosensors

    • Co-express fluorescently tagged MTMR2 with PI(3)P sensors (e.g., 2xFYVE-GFP)

    • Conduct time-lapse imaging to track changes in endosomal PI(3)P levels

    • Quantify correlation between MTMR2 recruitment and PI(3)P depletion

  • Endosomal subtype markers co-localization

    • Express wild-type or phosphorylation-site mutants of MTMR2 (S58A, S631A equivalents)

    • Co-stain for different endosomal subtypes (Rab5 for early endosomes, APPL1 for signaling endosomes, Rab7 for late endosomes)

    • Quantify changes in MTMR2 distribution between endosomal populations

  • Super-resolution microscopy

    • Use techniques like STORM or PALM to visualize nanoscale organization of MTMR2 on endosomal membranes

    • Co-localize with cargo proteins to assess effects on sorting

Functional approaches:

  • Cargo trafficking assays

    • Track fluorescently labeled endocytic cargo (e.g., transferrin, EGF)

    • Evaluate rates of internalization, recycling, and degradation in cells with manipulated MTMR2 expression

    • Compare effects of wild-type versus catalytically inactive MTMR2

  • Endosomal signaling studies

    • Monitor activation of signaling pathways initiated from endosomes (e.g., ERK1/2)

    • Expression of phosphorylation-deficient MTMR2 (S58A) has been shown to increase ERK1/2 activation

    • Use phospho-specific antibodies to quantify signaling outputs

  • Endosomal maturation kinetics

    • Track conversion of Rab5-positive to Rab7-positive endosomes over time

    • Evaluate effects of MTMR2 manipulation on maturation rates

    • Correlate with changes in phosphoinositide composition

Advanced techniques:

  • Endosomal isolation and proteomics

    • Isolate endosomal fractions from cells with manipulated MTMR2 expression

    • Perform proteomic analysis to identify changes in endosomal protein composition

    • Correlate with alterations in phosphoinositide levels

  • In vitro endosomal reconstitution

    • Generate artificial endosomal membranes with defined phosphoinositide composition

    • Add purified MTMR2 and monitor effects on membrane recruitment of endosomal proteins

    • Assess impact on fusion and fission events

The following table summarizes expected effects of different MTMR2 variants on endosomal properties:

MTMR2 VariantEndosomal LocalizationPI(3)P LevelsERK1/2 ActivationEndosomal Maturation
Wild-typePrimarily cytoplasmicNormalBaselineNormal rate
Catalytically inactive (C417S)Similar to wild-typeNormalBaselineNormal rate
S58ARab5-positive endosomesDecreasedIncreasedPotentially delayed
S58A/S631AAPPL1-positive endosomesSignificantly decreasedStrongly increasedSignificantly delayed

What methodologies can effectively compare chicken MTMR2 with its human ortholog for translational research?

Translational research comparing chicken MTMR2 with its human ortholog requires methodologies that address structural, functional, and regulatory aspects of these proteins. The following approaches enable comprehensive comparative analysis:

Structural comparison approaches:

  • Sequence and phylogenetic analysis

    • Perform multiple sequence alignment of MTMR2 from chicken, human, and other species

    • Calculate sequence identity/similarity percentages for full-length proteins and individual domains

    • Generate phylogenetic trees to visualize evolutionary relationships

  • 3D structural comparison

    • Generate homology models of chicken MTMR2 based on existing crystal structures

    • Compare with human MTMR2 structural data

    • Identify conserved and divergent regions in catalytic sites and interaction interfaces

  • Protein-protein interaction domain analysis

    • Compare binding motifs, particularly the C-terminal PDZ-binding motif important for PSD-95 interaction

    • Validate conservation of binding partners experimentally

Functional comparison approaches:

  • Parallel enzymatic assays

    • Express and purify recombinant human and chicken MTMR2

    • Conduct side-by-side phosphatase assays using identical substrates and conditions

    • Compare kinetic parameters (Km, Vmax, kcat) and substrate preferences

  • Cellular complementation studies

    • Knockdown endogenous MTMR2 in relevant cell types

    • Rescue with either human or chicken MTMR2

    • Compare ability to restore normal phenotypes (endosomal morphology, PI(3)P levels, signaling)

  • Cross-species functional validation

    • Express fluorescently tagged chicken MTMR2 in human cells and vice versa

    • Assess localization, enzymatic activity, and binding partner interactions

    • Evaluate functional equivalence through phenotypic rescue experiments

Regulatory comparison approaches:

  • Phosphorylation site conservation analysis

    • Identify equivalent regulatory phosphorylation sites (Ser58, Ser631) in chicken MTMR2

    • Create corresponding phosphorylation site mutants

    • Compare effects on subcellular localization and function

  • Cross-species kinase assays

    • Test whether human kinases (e.g., ERK1/2) can phosphorylate chicken MTMR2 and vice versa

    • Compare phosphorylation efficiency and site specificity

  • Differential response to cellular stressors

    • Expose cells expressing either human or chicken MTMR2 to various stressors (osmotic stress, oxidative stress)

    • Compare changes in localization, phosphorylation state, and activity

Disease-relevant comparative studies:

  • CMT4B1-associated mutation effects

    • Introduce equivalent CMT4B1-associated mutations into chicken MTMR2

    • Compare effects on protein stability, localization, and function with human mutants

    • Evaluate species-specific differences in pathogenic mechanisms

  • Drug screening applications

    • Use both chicken and human MTMR2 in parallel drug screening assays

    • Identify compounds that modulate activity or localization

    • Compare species-specific responses to lead compounds

For partial chicken MTMR2 constructs, ensure that the comparison focuses on equivalent domains between species and clearly document which regions are being compared. This approach allows for meaningful translational insights while acknowledging limitations of the partial protein construct.

How can I troubleshoot expression and purification issues with recombinant chicken MTMR2?

Troubleshooting expression and purification challenges for recombinant chicken MTMR2 requires systematic analysis of potential issues. The following methodological approaches address common problems:

Low expression yield:

  • Codon optimization

    • Analyze chicken MTMR2 sequence for rare codons in the expression host

    • Use codon-optimized synthetic genes for the expression system

    • For E. coli expression, consider Rosetta strains that supply rare tRNAs

  • Expression construct design

    • Try different affinity tags (His6, GST, MBP) and tag positions (N-terminal vs. C-terminal)

    • For partial constructs, ensure proper start/stop codons and reading frame

    • Include TEV or PreScission protease sites for tag removal

  • Expression conditions optimization

    • Test different induction temperatures (16°C, 25°C, 37°C)

    • Vary inducer concentration (0.1-1.0 mM IPTG for bacterial systems)

    • Extend expression time (overnight at lower temperatures)

    • For mammalian expression, test different transfection reagents and cell densities

Poor solubility:

  • Buffer optimization

    • Screen different buffer conditions (pH 6.5-8.5)

    • Test various salt concentrations (150-500 mM NaCl)

    • Add solubility enhancers (5-10% glycerol, 0.1-1% Triton X-100, 1-5 mM EDTA)

  • Fusion partners

    • Use solubility-enhancing fusion tags (MBP, SUMO, TrxA)

    • For partial constructs, consider whether the fragment might expose hydrophobic regions

  • Co-expression strategies

    • Co-express with chaperones (GroEL/ES, DnaK/J) in bacterial systems

    • Consider co-expression with binding partners that may stabilize the protein

Protein degradation:

  • Protease inhibition

    • Use comprehensive protease inhibitor cocktails in all buffers

    • Add specific inhibitors based on observed degradation patterns

    • Reduce processing time and maintain samples at 4°C

  • Stability screening

    • Perform thermal shift assays to identify stabilizing buffer components

    • Test additives like ATP, metal ions, or substrate analogs that might stabilize the protein

  • Site-directed mutagenesis

    • Identify and mutate potential protease-sensitive sites

    • For partial constructs, consider adding stabilizing elements at truncation points

Low activity or misfolding:

  • Refolding protocols

    • If protein is in inclusion bodies, develop a refolding protocol using gradual dialysis

    • Test different redox conditions to ensure proper disulfide bond formation

  • Activity assay optimization

    • Ensure proper reaction conditions (pH, ionic strength, temperature)

    • Verify substrate quality and accessibility

    • Include positive controls with known phosphatase activity

  • Structural validation

    • Use circular dichroism to confirm secondary structure elements

    • Employ limited proteolysis to assess domain folding

    • Consider FTIR or NMR for more detailed structural information

For partial MTMR2 constructs, additional considerations include ensuring that domain boundaries are properly defined to avoid disrupting structural elements, and verifying that the construct contains all necessary catalytic residues if enzymatic activity is required.

What controls are essential when studying chicken MTMR2 phosphoinositide interactions?

Rigorous controls are critical when studying chicken MTMR2 phosphoinositide interactions to ensure reliable and interpretable results. The following controls should be incorporated into experimental designs:

Protein-related controls:

  • Catalytically inactive mutant

    • Generate the equivalent of the C417S mutation identified in mammalian MTMR2

    • Use as a negative control in phosphatase assays

    • Should bind but not hydrolyze phosphoinositide substrates

  • Phosphorylation site mutants

    • Create phosphomimetic (S→D/E) and phospho-deficient (S→A) mutations of key regulatory sites

    • Essential for validating the role of phosphorylation in localization and activity

    • Use in combination with phosphorylation site-specific antibodies

  • Domain deletion constructs

    • Generate constructs lacking specific domains (PH-GRAM domain, catalytic domain)

    • Use to validate domain-specific contributions to phosphoinositide binding and catalysis

Substrate-related controls:

  • Non-hydrolyzable substrate analogs

    • Use modified phosphoinositides resistant to hydrolysis

    • Help distinguish binding from catalytic effects

  • Competitive inhibitors

    • Include inositol polyphosphates as competitive inhibitors

    • Test specificity by comparing inhibition profiles with different phosphoinositides

  • Substrate specificity panel

    • Test activity against all phosphoinositide species (PI3P, PI(3,5)P2, PI(4,5)P2, etc.)

    • Include non-substrate phospholipids (phosphatidylcholine, phosphatidylserine) as negative controls

Assay-specific controls:

  • For in vitro phosphatase assays:

    • Include time zero measurements to establish baseline

    • Run parallel reactions without enzyme to control for spontaneous hydrolysis

    • Include known phosphatases with defined specificity as positive controls

    • Use phosphate standards for quantification

  • For binding assays:

    • Perform protein-only controls to assess non-specific binding

    • Include gradients of protein concentration to determine binding parameters

    • Use liposomes lacking phosphoinositides as negative controls

  • For cellular localization studies:

    • Co-express established phosphoinositide biosensors (2xFYVE-GFP for PI(3)P, ML1N for PI(3,5)P2)

    • Include treatments that alter phosphoinositide levels (wortmannin for PI3K inhibition)

    • Use phosphoinositide phosphatase inhibitors to validate catalytic effects

Methodology validation controls:

  • Independent technique validation

    • Confirm key findings using multiple methodologies (e.g., both TLC and malachite green assays)

    • Cross-validate binding using both liposome sedimentation and surface plasmon resonance

  • Kinase/phosphatase treatment controls

    • Treat purified protein with λ-phosphatase to remove phosphorylation

    • Use in vitro kinase assays to generate specifically phosphorylated protein

  • Lipid extraction and detection controls

    • Include internal standards for mass spectrometry-based lipidomics

    • Perform spike-in experiments to validate extraction efficiency

The following table summarizes essential controls for different experimental approaches:

Experimental ApproachEssential ControlsPurpose
In vitro phosphatase assaysCatalytically inactive mutant (C417S equivalent), No-enzyme control, Time-zero measurementValidate enzymatic activity and specificity
Phosphoinositide bindingPH-GRAM domain deletion, Non-phosphoinositide lipids, Concentration gradientDetermine binding specificity and affinity
Cellular localizationPhosphomimetic and phospho-deficient mutants, Phosphoinositide-modifying drug treatmentsEstablish regulation of localization
Functional rescue experimentsWild-type protein, Domain deletion mutants, Catalytically inactive mutantDetermine structure-function relationships

What strategies can address challenges in studying chicken MTMR2 in primary neuronal cultures?

Studying chicken MTMR2 in primary neuronal cultures presents unique challenges requiring specialized approaches. The following strategies address common difficulties:

Efficient gene delivery and expression:

  • Optimized nucleofection protocols

    • Use Amaxa Nucleofector with neuron-specific programs

    • Optimize DNA:lipid ratios for primary neuron transfection

    • Consider co-transfection with fluorescent markers at 1:3 ratio for cell identification

    • For MTMR2 knockdown studies, validated siRNA delivery has been achieved in DRG neurons with observable effects on Piezo2 function

  • Viral delivery systems

    • Use adeno-associated viruses (AAVs) with neuron-specific promoters

    • Consider lentiviral systems for stable expression in long-term cultures

    • For partial MTMR2 constructs, ensure proper subcellular targeting with appropriate signal sequences

  • Promoter selection

    • Use neuron-specific promoters (synapsin, CaMKII) for targeted expression

    • Consider inducible systems (Tet-On) for temporal control of expression

Physiologically relevant readouts:

  • Subcellular localization analysis

    • Co-stain with markers for neuronal compartments (MAP2 for dendrites, Tau for axons)

    • Use super-resolution microscopy for detailed endosomal localization

    • Perform live imaging with low-level expression to avoid overexpression artifacts

    • MTMR2 has been successfully visualized in dendrites of cultured neurons, with discrete clusters co-localizing with PSD-95 and synaptophysin, indicating presence at excitatory synapses

  • Functional assays

    • Electrophysiological recordings of synaptic transmission

    • Calcium imaging to monitor activity patterns

    • FM dye uptake/release for presynaptic function

    • Knockdown of MTMR2 in cultured hippocampal neurons has been shown to reduce excitatory synapse number and suppress synaptic transmission

  • Endosomal trafficking assessment

    • Track fluorescently labeled endocytic cargo

    • Monitor endosomal maturation using pulse-chase approaches

    • Quantify colocalization with endosomal markers over time

Technical challenges and solutions:

  • Low transfection efficiency

    • Implement sparse labeling approaches for single-cell analysis

    • Use FACS to enrich transfected populations where applicable

    • Consider ex vivo electroporation of intact ganglia before dissociation

  • Phototoxicity during imaging

    • Use oxygen scavengers in imaging buffers

    • Employ low-light imaging techniques (EM-CCD cameras, light sheet microscopy)

    • Minimize exposure times and frequency for live-cell experiments

  • Heterogeneous neuronal populations

    • Use cell type-specific markers to identify neuronal subtypes

    • Consider magnetic-activated cell sorting (MACS) for purification

    • Implement single-cell transcriptomics to correlate MTMR2 function with cell identity

Specialized approaches for chicken neurons:

  • Species-specific considerations

    • Use chicken-specific antibodies where available

    • Validate siRNA sequences against chicken MTMR2 sequence

    • Consider species-optimized culture media supplements

  • Developmental timing

    • Adjust culture protocols for chicken embryonic neurons

    • Consider appropriate developmental stages for specific experiments

    • Account for species differences in maturation rate

  • Combined approaches

    • Complement in vitro studies with ex vivo preparations (e.g., intact DRG)

    • Consider in ovo electroporation for developmental studies

    • Validate findings in multiple neuronal subtypes

For mechanosensory studies related to MTMR2-Piezo2 interaction, specialized approaches have been developed and validated. Researchers have successfully performed siRNA-mediated knockdown of MTMR2 in DRG cultures and measured changes in rapidly adapting mechanically activated (RA-MA) currents, demonstrating that decreased expression of MTMR2 potentiates these currents while increased expression suppresses them .

How can chicken MTMR2 research contribute to understanding neurodegenerative disorders?

Chicken MTMR2 research offers valuable insights into neurodegenerative disorders, particularly those involving defects in endosomal trafficking and phosphoinositide metabolism. The translational potential spans several important areas:

Charcot-Marie-Tooth disease type 4B1 (CMT4B1):

  • Comparative disease modeling

    • Mutations in the MTMR2 gene in Schwann cells lead to CMT4B1, a severe demyelinating peripheral neuropathy

    • Chicken models can provide evolutionary perspective on conserved pathogenic mechanisms

    • Comparing chicken and human MTMR2 function can identify conserved regulatory pathways essential for myelin maintenance

  • Therapeutic target validation

    • Test whether chicken MTMR2 with CMT4B1-equivalent mutations shows similar dysfunction

    • Use partial constructs containing specific domains to identify which regions are critical for disease pathology

    • Evaluate whether rescuing phosphoinositide balance through alternative pathways can compensate for MTMR2 dysfunction

  • Schwann cell-neuron interaction studies

    • Examine how MTMR2 regulates communication between axons and myelinating cells

    • Assess how alterations in PI(3)P and PI(3,5)P2 levels affect myelin stability

    • Compare evolutionary conservation of these interactions across species

Synaptic dysfunction in neurodegenerative conditions:

  • Excitatory synapse regulation

    • MTMR2 localizes to excitatory synapses through interaction with PSD-95

    • Knockdown of MTMR2 in neurons reduces excitatory synapse density and function

    • These findings suggest potential roles in synaptic pathologies associated with various neurodegenerative disorders

  • Endosomal signaling pathways

    • MTMR2 regulates ERK1/2 activation through control of endosomal phosphoinositides

    • ERK1/2 dysregulation is implicated in numerous neurodegenerative diseases

    • Comparative studies between species can identify conserved signaling nodes as potential therapeutic targets

  • Protein trafficking defects

    • MTMR2 influences endosomal morphology and function

    • Many neurodegenerative diseases feature defective protein trafficking

    • Chicken neurons can provide a comparative system to study evolutionary conservation of trafficking mechanisms

Mechanosensory disorders:

  • MTMR2-Piezo2 interaction

    • MTMR2 regulates Piezo2-mediated mechanosensation through PI(3,5)P2 modulation

    • This pathway may be relevant for tactile dysfunction in peripheral neuropathies

    • Comparative studies can identify species-specific adaptations in mechanosensory regulation

  • Pain processing modulation

    • Investigating whether MTMR2-dependent regulation of sensory channels contributes to neuropathic pain

    • Testing if targeting this pathway could provide novel analgesic approaches

    • Avian models provide evolutionary perspective on conserved nociceptive mechanisms

Research approaches with translational potential:

  • Phosphoinositide-targeted therapeutics

    • Develop compounds that normalize phosphoinositide imbalances in MTMR2-deficient cells

    • Test effects on neuronal function and myelination

    • Compare efficacy across species to identify evolutionarily conserved mechanisms

  • Gene therapy strategies

    • Evaluate viral-mediated delivery of functional MTMR2 to rescue defects

    • Assess whether partial MTMR2 constructs containing only essential domains can provide therapeutic benefit

    • Compare outcomes in chicken and mammalian models

  • Biomarker development

    • Identify changes in phosphoinositide profiles that could serve as diagnostic markers

    • Develop assays for MTMR2 activity/phosphorylation as disease progression indicators

    • Validate conservation of these markers across species

The comparative study of chicken and mammalian MTMR2 can reveal which aspects of MTMR2 function are fundamentally conserved across evolution and therefore likely critical for basic neuronal health versus those that may represent species-specific adaptations.

What are promising directions for investigating MTMR2's role in endosomal signaling networks?

MTMR2's position at the intersection of phosphoinositide metabolism and endosomal function makes it a key regulator of cellular signaling networks. Several promising research directions can advance our understanding of these complex interactions:

Integration of phosphoinositide metabolism with signal transduction:

  • MAPK pathway regulation

    • Evidence indicates that MTMR2 regulation of endosomal PI(3)P and PI(3,5)P2 affects ERK1/2 activation

    • ERK1/2 in turn phosphorylates MTMR2 at Ser58, creating a feedback loop

    • Future research can explore how this feedback circuit responds to different stimuli

    • Comparative studies with chicken MTMR2 can reveal evolutionary conservation of this regulatory mechanism

  • Receptor tyrosine kinase (RTK) trafficking

    • Investigate how MTMR2-mediated phosphoinositide modulation affects internalization, recycling, and degradation of RTKs

    • Examine whether MTMR2 differentially regulates distinct RTK family members

    • Study how growth factor-induced signaling cascades are shaped by MTMR2 activity

  • Interplay with mTOR signaling

    • PI(3,5)P2 regulates mTORC1 activity on lysosomes

    • Explore whether MTMR2 modulation of PI(3,5)P2 affects mTOR-dependent growth and metabolism pathways

    • Investigate potential implications for diseases with dysregulated mTOR signaling

Spatiotemporal regulation of endosomal maturation:

  • Endosomal subtype transitions

    • Research indicates that MTMR2 differentially localizes to APPL1-positive versus Rab5-positive endosomes based on its phosphorylation state

    • Future studies can explore how this differential targeting affects signaling outcomes

    • Live imaging with phosphoinositide biosensors can reveal dynamic changes during endosomal maturation

  • Cargo-specific sorting decisions

    • Investigate whether MTMR2 differentially affects trafficking of distinct cargo types

    • Examine potential cargo-dependent recruitment of MTMR2 to specific endosomal subpopulations

    • Study how these sorting decisions impact downstream signaling events

  • Membrane contact sites

    • Explore MTMR2's potential role at endosome-ER contact sites

    • Examine how phosphoinositide conversion at these interfaces regulates calcium signaling and lipid transfer

    • Investigate protein complexes that may coordinate MTMR2 activity at membrane contact sites

Systems biology approaches:

  • Quantitative phosphoproteomics

    • Perform global phosphoproteomic analysis in cells with manipulated MTMR2 expression/activity

    • Identify signaling networks affected by MTMR2-dependent phosphoinositide changes

    • Compare phosphoproteomic patterns between wild-type and phosphorylation site mutants

  • Spatial proteomics of endosomal subpopulations

    • Isolate different endosomal subtypes and perform proteomic analysis

    • Compare protein composition in control versus MTMR2-depleted conditions

    • Identify proteins whose endosomal association is regulated by MTMR2-dependent phosphoinositide balance

  • Mathematical modeling of feedback circuits

    • Develop computational models of the MTMR2-ERK1/2 feedback loop

    • Simulate system behavior under various perturbations

    • Predict interventions that might normalize signaling in disease states

Methodological innovations:

  • Optogenetic control of MTMR2 localization

    • Develop light-inducible recruitment systems to target MTMR2 to specific endosomal populations

    • Study acute effects of localized phosphoinositide conversion

    • Examine how temporal dynamics of MTMR2 recruitment shape signaling outcomes

  • Biosensors for MTMR2 conformational states

    • Design FRET-based sensors to detect MTMR2 activation/inhibition

    • Monitor real-time changes in MTMR2 activity in response to cellular stimuli

    • Correlate with phosphoinositide dynamics and downstream signaling events

  • Single-endosome analysis

    • Develop methods to isolate and analyze individual endosomes

    • Characterize heterogeneity in phosphoinositide composition and MTMR2 association

    • Correlate with functional outcomes like signaling activity or maturation fate

The table below summarizes key aspects of MTMR2 involvement in endosomal signaling and promising research directions:

Signaling AspectCurrent KnowledgeFuture Research Directions
ERK1/2 pathwayMTMR2 phosphorylation by ERK1/2 regulates localization; MTMR2 activity affects ERK1/2 activationCharacterize feedback dynamics; identify additional components in the regulatory circuit
Endosomal subtype targetingPhosphorylation status determines localization to Rab5 vs. APPL1 endosomesDetermine mechanisms of differential recruitment; identify additional regulatory phosphorylation sites
Piezo2 regulationMTMR2 attenuates Piezo2 activity through PI(3,5)P2 depletionExplore physiological contexts of this regulation; investigate potential role in mechanosensory adaptation
Synaptic functionMTMR2 regulates excitatory synapse density and functionElucidate detailed mechanisms of synaptic maintenance; investigate activity-dependent regulation

What novel techniques could advance our understanding of MTMR2 function in phosphoinositide regulation?

Advancing our understanding of MTMR2 function in phosphoinositide regulation requires innovative methodological approaches that can capture the dynamic, spatiotemporally regulated nature of phosphoinositide metabolism. The following novel techniques hold particular promise:

Advanced imaging approaches:

  • Super-resolution phosphoinositide imaging

    • Implement STORM, PALM, or STED microscopy with phosphoinositide-specific probes

    • Enable nanoscale visualization of phosphoinositide domains on endosomal membranes

    • Track MTMR2-mediated changes in phosphoinositide distribution at unprecedented resolution

  • Single-molecule tracking of MTMR2

    • Use photoactivatable fluorescent proteins fused to MTMR2

    • Track individual MTMR2 molecules on endosomal surfaces

    • Analyze dwell times, diffusion coefficients, and interaction dynamics

  • FRET-based activity sensors

    • Develop intramolecular FRET sensors for MTMR2 conformational changes

    • Create intermolecular FRET pairs between MTMR2 and its substrates

    • Monitor real-time enzymatic activity in living cells

Genome engineering approaches:

  • Endogenous tagging via CRISPR-Cas9

    • Insert fluorescent tags into the endogenous MTMR2 locus

    • Maintain physiological expression levels and regulation

    • Compare localization and dynamics with overexpression systems

  • Phosphoinositide manipulation tools

    • Develop CRISPR-based methods to modify phosphoinositide-metabolizing enzymes

    • Create cell lines with altered phosphoinositide homeostasis

    • Test how system-wide phosphoinositide changes affect MTMR2 function

  • Rapid protein degradation systems

    • Implement auxin-inducible or dTAG degron systems for MTMR2

    • Study acute effects of MTMR2 depletion on phosphoinositide balance

    • Analyze temporal aspects of compensatory mechanisms

Biochemical and biophysical innovations:

  • Phosphoinositide microarrays

    • Generate arrays with defined phosphoinositide compositions

    • Test binding of wild-type versus mutant MTMR2

    • Identify co-factors that modify binding or activity

  • Reconstituted endosomal systems

    • Create synthetic endosomal membranes with defined lipid composition

    • Add purified MTMR2 and monitor enzymatic activity

    • Incorporate additional proteins to study regulatory interactions

  • Hydrogen-deuterium exchange mass spectrometry

    • Map conformational changes in MTMR2 upon substrate binding or protein interaction

    • Identify allosteric regulatory sites

    • Compare structural dynamics between wild-type and disease-associated mutants

Spatial proteomics and lipidomics:

  • Proximity labeling of MTMR2 microenvironments

    • Use BioID or APEX2 fusions to MTMR2

    • Identify proteins in proximity to MTMR2 on different endosomal subtypes

    • Compare interactomes based on phosphorylation state

  • Subcellular phosphoinositide lipidomics

    • Isolate distinct endosomal populations

    • Perform targeted mass spectrometry to quantify phosphoinositide species

    • Compare profiles between control and MTMR2-manipulated conditions

  • Spatial transcriptomics correlation

    • Analyze transcriptional responses to MTMR2 manipulation

    • Correlate with phosphoinositide changes and signaling outputs

    • Identify gene expression signatures of altered endosomal function

Computational and systems approaches:

  • Molecular dynamics simulations

    • Model MTMR2 interactions with membrane phosphoinositides

    • Simulate effects of phosphorylation on protein conformation and membrane association

    • Predict effects of disease-associated mutations on catalytic function

  • Network analysis of phosphoinositide-dependent signaling

    • Integrate proteomic, lipidomic, and phosphoproteomic data

    • Identify signaling nodes regulated by MTMR2-dependent phosphoinositide changes

    • Generate testable predictions about system behavior

  • Machine learning image analysis

    • Train algorithms to identify and classify endosomal subtypes

    • Quantify subtle changes in morphology and distribution

    • Extract features not obvious to human observers

The table below summarizes how these novel techniques could address specific aspects of MTMR2 function:

Aspect of MTMR2 FunctionTraditional ApproachesNovel TechniquesPotential Insights
Substrate specificityIn vitro phosphatase assaysPhosphoinositide microarrays; reconstituted membranesFine-tuned preferences for membrane contexts; influence of curvature and lipid packing
Phosphorylation regulationPhosphomimetic mutationsOptogenetic kinase control; phospho-specific intramolecular sensorsReal-time visualization of phosphorylation-dependent localization; temporal dynamics of feedback loops
Endosomal targetingColocalization with markersSingle-molecule tracking; super-resolution imagingNanoscale organization on endosomal membranes; relationship to endosomal maturation state
Protein-protein interactionsCo-immunoprecipitation; Y2HProximity labeling; quantitative interactomicsContext-dependent interaction networks; temporal dynamics of complex formation

For partial MTMR2 constructs, these novel techniques can provide valuable insights into domain-specific functions and interactions, helping to delineate which protein regions are responsible for particular aspects of MTMR2 biology.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.